Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Nanotechnology ; 33(8)2021 Dec 02.
Article in English | MEDLINE | ID: mdl-34261054

ABSTRACT

Bcl-2, an anti-apoptotic protein, is always overexpressed in tumor cells to suppress the pro-apoptotic function of Bax, thereby prolonging the life of the tumor. However, BH3 proteins could directly activate Bax via antagonizing Bcl-2 to induce apoptosis in response to the stimulation. Thus, mimicking BH3 proteins with a peptide is a potential strategy for anti-cancer therapy. Unfortunately, clinical translation of BH3-mimic peptide is hindered by its inefficacious cellular internalization and proteolysis resistance. Herein, we translated a BH3-mimic peptide into a peptide-auric spheroidal nanocluster (BH3-AuNp), in which polymeric BH3-Auric precursors [Au1+-S-BH3]narein situself-assembled on the surface of gold nanoparticles by a one-pot synthesis. Expectedly, this strategy could improve the anti-proteolytic ability and cytomembrane penetrability of the BH3 peptide. As a result, BH3-AuNp successfully induced the apoptosis of two cancer cell lines by an order of magnitude compared to BH3. This therapeutic and feasible peptide nano-engineering strategy will help peptides overcome the pharmaceutical obstacles, awaken its biological functions, and possibly revive the research about peptide-derived nanomedicine.


Subject(s)
Apoptosis/drug effects , Drug Delivery Systems/methods , Metal Nanoparticles/chemistry , Peptide Fragments , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Survival/drug effects , Gold/chemistry , HCT116 Cells , Humans , Nanomedicine , Neoplasms/chemistry , Neoplasms/metabolism , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/pharmacokinetics , Proto-Oncogene Proteins/pharmacology
2.
ACS Chem Biol ; 10(9): 2149-57, 2015 Sep 18.
Article in English | MEDLINE | ID: mdl-26151238

ABSTRACT

Hydrocarbon stapling has been applied to restore and stabilize the α-helical structure of bioactive peptides for biochemical, structural, cellular, and in vivo studies. The peptide sequence, in addition to the composition and location of the installed staple, can dramatically influence the properties of stapled peptides. As a result, constructs that appear similar can have distinct functions and utilities. Here, we perform a side-by-side comparison of stapled peptides modeled after the pro-apoptotic BIM BH3 helix to highlight these principles. We confirm that replacing a salt-bridge with an i, i + 4 hydrocarbon staple does not impair target binding affinity and instead can yield a biologically and pharmacologically enhanced α-helical peptide ligand. Importantly, we demonstrate by electron microscopy that the pro-apoptotic activity of a stapled BIM BH3 helix correlates with its capacity to achieve cellular uptake without membrane disruption and accumulate at the organellar site of mechanistic activity.


Subject(s)
Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/pharmacology , Apoptosis/drug effects , Hydrocarbons/chemistry , Hydrocarbons/pharmacology , Membrane Proteins/chemistry , Membrane Proteins/pharmacology , Peptides/chemistry , Peptides/pharmacology , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/pharmacology , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/pharmacokinetics , Bcl-2-Like Protein 11 , Cell Line , Hydrocarbons/pharmacokinetics , Membrane Proteins/pharmacokinetics , Mice , Molecular Sequence Data , Peptides/pharmacokinetics , Protein Structure, Secondary , Protein Structure, Tertiary , Proto-Oncogene Proteins/pharmacokinetics
3.
Histochem Cell Biol ; 141(1): 111-21, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24141902

ABSTRACT

The localization of DEAD (Asp-Glu-Ala-Asp) box helicase 6 (DDX6) in spermatogenic cells from the mouse, rat, and guinea pig was studied by immunofluorescence (IF) and immunoelectron microscopy (IEM). Spermatogenic cells from these species yielded similar DDX6 localization pattern. IF microscopy results showed that DDX6 localizes to both the nucleus and cytoplasm. In the cytoplasm of spermatogenic cells, diffuse cytosolic and discrete granular staining was observed, with the staining pattern changing during cell differentiation. IEM revealed that DDX6 localized to the five different types of nuage structures and non-nuage structures, including small granule aggregate and late spermatid annuli. Nuclear labeling was strongest in leptotene and zygotene spermatocytes and moderately strong in the nuclear pocket of late spermatids. DDX6 also localized to the surface of outer dense fibers, which comprise of flagella. The results show that DDX6 is present in nuage and non-nuage structures as well as nuclei, suggesting that DDX6 has diverse functions in spermatogenic cells.


Subject(s)
DEAD-box RNA Helicases/pharmacokinetics , Proto-Oncogene Proteins/pharmacokinetics , RNA Nucleotidyltransferases/pharmacokinetics , Spermatogenesis/physiology , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , Fluorescent Antibody Technique , Guinea Pigs , Male , Mice , Microscopy, Immunoelectron , Rats , Rats, Wistar , Spermatids/cytology , Spermatocytes/cytology , Spermatogonia/cytology , Testis/cytology , Testis/embryology
4.
J Mol Biol ; 425(10): 1655-69, 2013 May 27.
Article in English | MEDLINE | ID: mdl-23416556

ABSTRACT

Members of the ETS family of transcription factors regulate a functionally diverse array of genes. All ETS proteins share a structurally conserved but sequence-divergent DNA-binding domain, known as the ETS domain. Although the structure and thermodynamics of the ETS-DNA complexes are well known, little is known about the kinetics of sequence recognition, a facet that offers potential insight into its molecular mechanism. We have characterized DNA binding by the ETS domain of PU.1 by biosensor-surface plasmon resonance (SPR). SPR analysis revealed a striking kinetic profile for DNA binding by the PU.1 ETS domain. At low salt concentrations, it binds high-affinity cognate DNA with a very slow association rate constant (≤10(5)M(-)(1)s(-)(1)), compensated by a correspondingly small dissociation rate constant. The kinetics are strongly salt dependent but mutually balance to produce a relatively weak dependence in the equilibrium constant. This profile contrasts sharply with reported data for other ETS domains (e.g., Ets-1, TEL) for which high-affinity binding is driven by rapid association (>10(7)M(-)(1)s(-)(1)). We interpret this difference in terms of the hydration properties of ETS-DNA binding and propose that at least two mechanisms of sequence recognition are employed by this family of DNA-binding domain. Additionally, we use SPR to demonstrate the potential for pharmacological inhibition of sequence-specific ETS-DNA binding, using the minor groove-binding distamycin as a model compound. Our work establishes SPR as a valuable technique for extending our understanding of the molecular mechanisms of ETS-DNA interactions as well as developing potential small-molecule agents for biotechnological and therapeutic purposes.


Subject(s)
DNA-Binding Proteins/pharmacokinetics , DNA/chemistry , DNA/pharmacokinetics , Proto-Oncogene Proteins/pharmacokinetics , Trans-Activators/pharmacokinetics , Animals , Base Sequence , Binding Sites/genetics , Binding, Competitive , Biosensing Techniques/methods , DNA/genetics , DNA-Binding Proteins/genetics , Distamycins/pharmacology , Mice , Models, Molecular , Protein Structure, Tertiary/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Static Electricity , Surface Plasmon Resonance/methods , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics
5.
Int J Biol Macromol ; 50(3): 796-814, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22155216

ABSTRACT

Bcl-2 (B cell lymphoma-2) is an anti-apoptotic member of Bcl-2 family and its overexpression causes development of several types of cancer. The BH3 domain of pro-apoptotic and BH3-only proteins is capable of binding to Bcl-2 protein to induce apoptosis. This binding is the basis for the development of novel anticancer drug which would likely antagonize Bcl-2 overexpression. In this study we have identified BH3 domain of Bax (Bax BH3) as potentially the best Bcl-2 antagonist by performing docking of BH3 peptides (peptides representing BH3 domain of pro-apoptotic and BH3-only proteins) into the Bcl-2 hydrophobic groove formed by BH3, BH1 and BH2 domains (also referred as BH3 cleft). To predict the best small antagonist for Bcl-2, three groups of small peptides (pentapeptide, tetrapeptide and tripeptide) were designed and screened against Bcl-2 which revealed the structural importance of a set of residues playing a vital role in interaction with Bcl-2. The docking and scoring function identified KRIG and KRI as specific peptides among the screened small peptides responsible for Bcl-2 neutralization and would induce apoptosis. The applied pharmacokinetic and pharmacological filters to all small peptides signify that only IGD has drug-like properties and displayed good oral bioavailability. However, the obtained binding affinity of IGD to Bcl-2 was diminutive. Hence deprotonation, amidation, acetylation, benzoylation, benzylation, and addition of phenyl, deoxyglucose and glucose fragments were performed to increase the binding affinity and to prevent its rapid degradation. Benzoylated IGD tripeptide (IGD(bzo)) was observed to have increased binding affinity than IGD with acceptable pharmacokinetic filters. In addition, stability of Bcl-2/IGD(bzo) complex was validated by Molecular Dynamics (MD) simulations revealing improved binding energy, salt bridges and strong interaction energies. This study suggests a new molecule that inhibits Bcl-2 associated cancer/tumor regression.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Neoplasms/drug therapy , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins/pharmacology , Amino Acid Sequence , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Biological Availability , Drug Screening Assays, Antitumor , Humans , Molecular Dynamics Simulation , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Peptide Fragments/therapeutic use , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/pharmacokinetics , Proto-Oncogene Proteins/therapeutic use , Proto-Oncogene Proteins c-bcl-2/chemistry , Thermodynamics
6.
J Neurochem ; 94(2): 502-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15998300

ABSTRACT

Recent studies have indicated that various growth factors are involved in synaptic functions; however, the precise mechanisms remain unclear. In order to elucidate the molecular mechanisms of the growth factor-mediated regulation of presynaptic functions, the effects of epidermal growth factor (EGF) and insulin-like growth factor-1 (IGF-1) on neurotransmitter release were studied in rat PC12 cells. Brief treatment with EGF and IGF-1 enhanced Ca2+-dependent dopamine release in a concentration-dependent manner. EGF activated both mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3-kinase) pathways, and the EGF-dependent enhancement of DA release was suppressed by a MAPK kinase inhibitor as well as by PI3-kinase inhibitors. In striking contrast, IGF-1 activated the PI3-kinase pathway but not the MAPK pathway, and IGF-1-dependent enhancement was suppressed by a PI3-kinase inhibitor but not by a MAPK kinase inhibitor. The enhanced green fluorescent protein-tagged pleckstrin homology (PH) domain of protein kinase B, which selectively binds to phosphatidylinositol 3,4-bisphosphate and phosphatidylinositol 3,4,5-triphosphate, was translocated to the plasma membrane after treatment with either EGF or NGF. By contrast, no significant redistribution was induced by IGF-1. These results indicate that PI3-kinase participates in the enhancement of neurotransmitter release by two distinct mechanisms: EGF and NGF activate PI3-kinase in the plasma membrane, whereas IGF-1 activates PI3-kinase possibly in the intracellular membrane, leading to enhancement of neurotransmitter release in a MAPK-dependent and -independent manner respectively.


Subject(s)
Cell Membrane/metabolism , Dopamine/metabolism , Extracellular Space/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Blotting, Western/methods , Calcium/metabolism , Cell Membrane/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epidermal Growth Factor/pharmacology , Extracellular Space/drug effects , Green Fluorescent Proteins/metabolism , Green Fluorescent Proteins/pharmacokinetics , Insulin-Like Growth Factor I/pharmacology , Ionomycin/pharmacology , Ionophores/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , PC12 Cells/drug effects , PC12 Cells/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/pharmacokinetics , Protein Transport/drug effects , Proto-Oncogene Proteins/pharmacokinetics , Proto-Oncogene Proteins c-akt , Rats , Time Factors
7.
Cancer Lett ; 223(2): 339-47, 2005 Jun 08.
Article in English | MEDLINE | ID: mdl-15896469

ABSTRACT

Wnt/beta-catenin signalling pathway is integrally associated with human tumour development and progression. Aberrant beta-catenin intracellular distribution has been found in gastric cancer, but the pattern of Wnt expression in stepwise gastrocarcinogenesis and its potential influence in beta-catenin distribution are still lesser known. By the methods of frozen tissue array-based immunohistochemistry, Western blot analysis and RT-PCR, a paralleled study was conducted to check Wnt2 expression and beta-catenin intracellular distribution in two major subtypes of gastric cancers (intestinal gastric cancer, i-GC and diffuse gastric cancer, d-GC) and their premalignant (intestinal metaplasia, IM and chronic gastritis, CG) and noncancerous counterparts. According to the results obtained and the clinical data collected, correlation of Wnt2 expression with beta-catenin translocalisation and their links with tumour dissemination were elucidated. The results demonstrated (1) that Wnt2 expression and cytoplasmic/nuclear beta-catenin accumulations appeared in most gastric cancers irrespective to their morphological phenotypes, (2) that over-expressed Wnt and nuclear translocalisation of beta-catenin were found in 68 and 58% of i-GCs and in 47 and 47% of d-GCs in a closely related pattern (P<0.01) and (3) that co-existence of Wnt2 up-regulation/beta-catenin nuclear translocalisation were positively associated with lymph node metastasis (P<0.05) as well as T-stage. These data indicate that Wnt/beta-catenin signalling pathway is activated in most of gastric cancers, which may play pivotal roles either in gastric cancer formation or in tumour invasion and dissemination.


Subject(s)
Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/pharmacokinetics , Gene Expression Profiling , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/pharmacokinetics , Stomach Neoplasms/genetics , Stomach Neoplasms/physiopathology , Trans-Activators/biosynthesis , Trans-Activators/pharmacokinetics , Blotting, Western , Cell Nucleus/chemistry , Cell Transformation, Neoplastic , China , Cytoplasm , Humans , Immunohistochemistry , Neoplasm Invasiveness , Neoplasm Metastasis , Precancerous Conditions , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Up-Regulation , Wnt2 Protein , beta Catenin
8.
Oncogene ; 24(15): 2574-9, 2005 Apr 07.
Article in English | MEDLINE | ID: mdl-15735705

ABSTRACT

Mdm2 and MdmX function as cellular regulators of the p53 tumor suppressor protein. Intriguingly, the activities of these proteins are interdependent; MdmX stabilizes Mdm2, enabling its activities towards p53, but it also requires Mdm2 for its nuclear localization. Here we demonstrate that via its phosphorylation by CDK2/Cdc2p34, MdmX regulates nuclear export of Mdm2. Cdc2p34 phosphorylates MdmX on Ser 96 in vitro. Mutation within this site (MdmX(S96A)) impairs, whereas phosphomimic substitution (MdmX(S96D)) increases the cytoplasmic localization of MdmX, suggesting that CDK2/Cdc2p34 phosphorylation is required for export of MdmX from the nucleus. Consequently, cells that express MdmX(S96A) retain Mdm2 in their nuclei, suggesting that export of Mdm2 to the cytoplasm is MdmX-dependent. Similarly, treatment of cells with the pharmacological inhibitor of CDK2/Cdc2p34 or with a dominant-negative Cdc2 results in nuclear localization of MdmX and Mdm2 and decreases the level of Mdm2 expression. Since Cdc2p34 is active in nonstressed conditions, our finding provides a novel insight into the signaling cascade involved in the regulation of MdmX localization and for regulation of Mdm2 localization and stability.


Subject(s)
CDC2-CDC28 Kinases/pharmacology , Nuclear Proteins/metabolism , Nuclear Proteins/pharmacokinetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/pharmacokinetics , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Cyclin-Dependent Kinase 2 , Cytoplasm , Humans , Phosphorylation , Proto-Oncogene Proteins c-mdm2 , Signal Transduction
9.
Brain Res Mol Brain Res ; 110(1): 15-26, 2003 Jan 31.
Article in English | MEDLINE | ID: mdl-12573529

ABSTRACT

Heterotrimeric GTP-binding proteins (G proteins) are involved in the coupling of a variety of cell surface receptors to different intracellular signalling pathways, some of which take part in the regulation of growth by affecting cell proliferation and/or differentiation. In cultured astrocytes, many receptors of neuropeptides and hormones are coupled to the heterotrimeric G(i) proteins which regulate the mitogen-activated protein kinase (MAPK/ERK) cascade through both the Galpha and Gbetagamma subunits. We have previously reported that functionally active recombinant myr-G(i2)alpha subunits added to such cultures are internalised and distributed within the plasma membrane and cytosol as well as in the nuclei of dividing astrocytes. Here we show that astrocytes proliferate dose-dependently in response to exogenous myr-G(i2)alpha subunits. Concentrations of 100 pM-30 nM myr-G(i2)alpha caused more than 2.5-fold increase of [3H]thymidine incorporation over basal levels. Other classes of myr-Galpha subunits, such as G(i3)alpha or G(o)alpha, induced a much lower proliferative effect. The addition of G(i1)alpha subunits to the cultures produced no change, indicating the selectivity of this effect. Even though myr-G(i2)alpha subunits are internalised by the cells regardless of their guanine nucleotide-bound state, much less [3H]thymidine incorporation was observed in the presence of GDPbetaS-myr-G(i2)alpha or GTPgammaS-myr-G(i2)alpha. Further, the fluorescent labelling was dissimilarly distributed, the signal being concentrated in the nucleus and perinuclear regions of the astrocytes. Selective disassembly of caveolae impaired both myr-G(i2)alpha internalisation and DNA induction. Together, these data reveal a proliferative effect of myr-G(i2)alpha subunits in astrocytes, and provide evidence for the incorporation of exogenous myr-G(i2)alpha subunits into the mitogen cascade activated by neurotransmitters or growth factors. The fact that Galpha proteins can enter cells is particularly interesting because options for delivering functional proteins into cells are limited. Thus, these proteins may have clinical applications for compensating deficits in the transduction mechanisms associated with several neurological diseases, or as a non-invasive membrane traversing carriers.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Fatty Acids, Monounsaturated/pharmacokinetics , GTP-Binding Protein alpha Subunits, Gi-Go/pharmacokinetics , Mitogens/pharmacokinetics , Proto-Oncogene Proteins/pharmacokinetics , Animals , Astrocytes/drug effects , Blood Proteins/pharmacology , Cell Division/drug effects , Cells, Cultured , Endocytosis/physiology , Fluorescent Dyes , GTP-Binding Protein alpha Subunit, Gi2 , Rats , Rats, Wistar
10.
Curr Opin Microbiol ; 5(1): 81-6, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11834374

ABSTRACT

The susceptibility of cystic fibrosis patients to bacterial pathogens is associated with deficient airway antimicrobial peptide activity, and airway-surface-liquid dehydration with decreased transport velocity and hypersecretion of mucus. Susceptibility to Pseudomonas aeruginosa infection has been linked to the role of the cystic fibrosis transmembrane conductance regulator protein as a receptor for P. aeruginosa. Binding of P. aeruginosa coordinates lung clearance as part of innate immunity. The function of CFTR in innate immunity to P. aeruginosa infection is multifactorial, with one key component being a specific ligand-receptor interaction between the protein and the microbe.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis/complications , Cystic Fibrosis/genetics , Lung/microbiology , Pseudomonas Infections/complications , Pseudomonas aeruginosa , Respiratory Tract Infections/microbiology , Animals , Apoptosis , Disease Susceptibility , Humans , Immunity, Innate , Models, Biological , Mutation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/pharmacokinetics , Proto-Oncogene Proteins c-fyn , Respiratory Tract Infections/genetics
11.
Cancer Res ; 60(22): 6253-8, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11103779

ABSTRACT

Growth of the human rhabdomyosarcoma A673 cell line in nude mice is substantially reduced but not completely suppressed after systemic administration of the antihuman vascular endothelial growth factor (VEGF) monoclonal antibody (Mab) A.4.6.1. Potentially, such escape might be attributable to incomplete local penetration of the antibody because of a diffusion barrier associated with tumor growth. Alternatively, it might reflect a compensatory up-regulation of murine VEGF, produced by the stroma of the host, or of other angiogenic factor genes. To test these potential mechanisms, systemic administration of Mab A.4.6.1, was performed in conjunction with intratumoral administration of an irrelevant antibody, an antihuman VEGF Fab or mFlt(1-3)-IgG that neutralizes both human and murine VEGF. Tumor growth in the systemic-plus-intratumoral anti-VEGF group was not different from that in the systemic anti-VEGF-plus-intratumoral-control antibody group, arguing against the possibility that bioavailability is the factor that limits the antitumor efficacy of Mab A.4.6.1. However, intratumoral mFlt(l-3)-IgG administration dramatically enhanced the activity of systemic anti-VEGF Mab and resulted in complete suppression of tumor growth, which indicated that host VEGF significantly contributes to tumor growth. Systemic administration of mFlt(1-3)-IgG alone replicated these findings. Histological analysis of residual tumor tissues revealed an almost complete absence of host-derived vasculature and massive tumor-cell necrosis in the mFlt(1-3)-IgG groups. Such extensive necrotic areas were not present in the other groups. Real-time reverse transcription-PCR analysis of total RNA derived from tumor tissues indicated strong up-regulation of both human and murine VEGF as well as other genes regulated by hypoxia. Our findings emphasize the need to completely block VEGF for maximal inhibition of tumor growth.


Subject(s)
Endothelial Growth Factors/antagonists & inhibitors , Lymphokines/antagonists & inhibitors , Neovascularization, Pathologic/pathology , Rhabdomyosarcoma/pathology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Biological Availability , Cell Division/physiology , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Endothelial Growth Factors/immunology , Gene Expression/drug effects , Humans , Immunoglobulin Fragments/immunology , Immunoglobulin Fragments/metabolism , Immunoglobulin Fragments/pharmacology , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Injections, Intralesional , Lymphokines/biosynthesis , Lymphokines/genetics , Lymphokines/immunology , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Proto-Oncogene Proteins/pharmacokinetics , Proto-Oncogene Proteins/pharmacology , RNA/genetics , RNA/metabolism , Receptor Protein-Tyrosine Kinases/pharmacokinetics , Receptor Protein-Tyrosine Kinases/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/blood supply , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors , Xenograft Model Antitumor Assays
12.
J Neurosci ; 19(23): 10348-56, 1999 Dec 01.
Article in English | MEDLINE | ID: mdl-10575032

ABSTRACT

Serotonin (5-HT) plays a crucial neuromodulatory role in numerous physiological and behavioral functions, and dysfunction of the serotonergic system has been implicated in several psychiatric disorders. Despite the widespread importance of the central serotonergic neurotransmitter system, little is known about the molecular mechanisms controlling the development of 5-HT neurons. We previously identified an ETS domain transcription factor, Pet-1, that is expressed in a small number of tissues, including the brain. Here, we show that expression of Pet-1 RNA in the brain is restricted to, and marks, the entire rostrocaudal extent of rat serotonergic hindbrain raphe nuclei. Remarkably, Pet-1 RNA colocalizes with tryptophan hydroxylase-positive neurons in raphe nuclei but not with their nonserotonergic neuron or non-neuronal neighbors. Pet-1 RNA is limited to two domains in the developing hindbrain, which precedes the appearance of 5-HT in each domain by approximately a half day. Conserved Pet-1 binding sites are present in or near the promoter regions of the human and mouse 5-HT1a receptor, serotonin transporter, tryptophan hydroxylase, and aromatic L-amino acid decarboxylase genes whose expression is characteristic of the serotonergic neuron phenotype. These sites are capable of supporting transcriptional activation through interactions with the Pet-1 ETS domain and can function as enhancers. Together, our findings establish Pet-1 as an early and precise marker of 5-HT neurons and suggest that it functions specifically in the differentiation and maintenance of these neurons.


Subject(s)
Brain/metabolism , Neurons/metabolism , Proto-Oncogene Proteins/pharmacokinetics , Serotonin/metabolism , Transcription Factors , Animals , Binding Sites/physiology , Biomarkers , Conserved Sequence/physiology , Embryo, Mammalian/physiology , Gene Expression , Neurons/enzymology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA/metabolism , Raphe Nuclei/cytology , Raphe Nuclei/enzymology , Raphe Nuclei/metabolism , Rats , Rats, Sprague-Dawley , Rhombencephalon/cytology , Rhombencephalon/embryology , Rhombencephalon/enzymology , Rhombencephalon/metabolism , Time Factors , Tyrosine 3-Monooxygenase/metabolism
13.
Biol Chem Hoppe Seyler ; 370(11): 1215-20, 1989 Nov.
Article in English | MEDLINE | ID: mdl-2692614

ABSTRACT

We have investigated the proteinase-inhibitory activity against cathepsin L of some c-Ha-ras gene products (p21s) with point mutations at position 12, 33 or 35, which affect the interaction with GTPase-activating protein (GAP). All of the full-length p21s examined showed similar inhibitory activities irrespective of the point mutation and the transforming activity. These results suggest that mutations at GAP-interacting sites have no effect on the proteinase-inhibitory activity of p21s and that the proteinase-inhibitory activity alone is not sufficient for the transformation caused by p21s.


Subject(s)
Cathepsins/antagonists & inhibitors , Cysteine Proteinase Inhibitors , Endopeptidases , Proto-Oncogene Proteins/pharmacokinetics , Amino Acid Sequence , Animals , Cathepsin L , Cysteine Endopeptidases , GTPase-Activating Proteins , Humans , Molecular Sequence Data , Mutation , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras) , Rats , Swine , ras GTPase-Activating Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...